Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
1.
J Intern Med ; 292(4): 667-678, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35670497

RESUMO

OBJECTIVE: Cross-sectional studies demonstrate that catecholamine stimulation of fat cell lipolysis is blunted in obesity. We investigated whether this defect persists after substantial weight loss has been induced by metabolic surgery, and whether it is related to the outcome. DESIGN/METHODS: Patients with obesity not able to successfully reduce body weight by conventional means (n = 126) were investigated before and 5 years after Roux-en-Y gastric bypass surgery (RYGB). They were compared with propensity-score matched subjects selected from a control group (n = 1017), and with the entire group after adjustment for age, sex, body mass index (BMI), fat cell volume and other clinical parameters. Catecholamine-stimulated lipolysis (glycerol release) was investigated in isolated fat cells using noradrenaline (natural hormone) or isoprenaline (synthetic beta-adrenoceptor agonist). RESULTS: Following RYGB, BMI was reduced from 39.9 (37.5-43.5) (median and interquartile range) to 29.5 (26.7-31.9) kg/m2 (p < 0.0001). The post-RYGB patients had about 50% lower lipolysis rates compared with the matched and total series of controls (p < 0.0005). Nordrenaline activation of lipolysis at baseline was associated with the RYGB effect; those with high lipolysis activation (upper tertile) lost 30%-45% more in body weight, BMI or fat mass than those with low (bottom tertile) initial lipolysis activation (p < 0.0007). CONCLUSION: Patients with obesity requiring metabolic surgery have impaired ability of catecholamines to stimulate lipolysis, which remains despite long-term normalization of body weight by RYGB. Furthermore, preoperative variations in the ability of catecholamines to activate lipolysis may predict the long-term reduction in body weight and fat mass.


Assuntos
Cirurgia Bariátrica , Derivação Gástrica , Obesidade Mórbida , Índice de Massa Corporal , Peso Corporal , Catecolaminas/farmacologia , Estudos Transversais , Glicerol , Hormônios , Humanos , Isoproterenol/farmacologia , Lipólise/fisiologia , Norepinefrina , Obesidade/metabolismo , Obesidade/cirurgia , Obesidade Mórbida/cirurgia , Receptores Adrenérgicos/metabolismo , Resultado do Tratamento
2.
Sci Rep ; 12(1): 3666, 2022 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-35256633

RESUMO

Total body fat and central fat distribution are heritable traits and well-established predictors of adverse metabolic outcomes. Lipolysis is the process responsible for the hydrolysis of triacylglycerols stored in adipocytes. To increase our understanding of the genetic regulation of body fat distribution and total body fat, we set out to determine if genetic variants associated with body mass index (BMI) or waist-hip-ratio adjusted for BMI (WHRadjBMI) in genome-wide association studies (GWAS) mediate their effect by influencing adipocyte lipolysis. We utilized data from the recent GWAS of spontaneous and isoprenaline-stimulated lipolysis in the unique GENetics of Adipocyte Lipolysis (GENiAL) cohort. GENiAL consists of 939 participants who have undergone abdominal subcutaneous adipose biopsy for the determination of spontaneous and isoprenaline-stimulated lipolysis in adipocytes. We report 11 BMI and 15 WHRadjBMI loci with SNPs displaying nominal association with lipolysis and allele-dependent gene expression in adipose tissue according to in silico analysis. Functional evaluation of candidate genes in these loci by small interfering RNAs (siRNA)-mediated knock-down in adipose-derived stem cells identified ZNF436 and NUP85 as intrinsic regulators of lipolysis consistent with the associations observed in the clinical cohorts. Furthermore, candidate genes in another BMI-locus (STX17) and two more WHRadjBMI loci (NID2, GGA3, GRB2) control lipolysis alone, or in conjunction with lipid storage, and may hereby be involved in genetic control of body fat. The findings expand our understanding of how genetic variants mediate their impact on the complex traits of fat storage and distribution.


Assuntos
Estudo de Associação Genômica Ampla , Lipólise , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Loci Gênicos , Humanos , Isoproterenol/metabolismo , Lipólise/genética , Fatores de Transcrição/metabolismo
3.
Diabetes ; 71(6): 1350-1362, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35320353

RESUMO

Interindividual differences in generation of new fat cells determine body fat and type 2 diabetes risk. In the GENetics of Adipocyte Lipolysis (GENiAL) cohort, which consists of participants who have undergone abdominal adipose biopsy, we performed a genome-wide association study (GWAS) of fat cell number (n = 896). Candidate genes from the genetic study were knocked down by siRNA in human adipose-derived stem cells. We report 318 single nucleotide polymorphisms (SNPs) and 17 genetic loci displaying suggestive (P < 1 × 10-5) association with fat cell number. Two loci pass threshold for GWAS significance, on chromosomes 2 (lead SNP rs149660479-G) and 7 (rs147389390-deletion). We filtered for fat cell number-associated SNPs (P < 1.00 × 10-5) using evidence of genotype-specific expression. Where this was observed we selected genes for follow-up investigation and hereby identified SPATS2L and KCTD18 as regulators of cell proliferation consistent with the genetic data. Furthermore, 30 reported type 2 diabetes-associated SNPs displayed nominal and consistent associations with fat cell number. In functional follow-up of candidate genes, RPL8, HSD17B12, and PEPD were identified as displaying effects on cell proliferation consistent with genetic association and gene expression findings. In conclusion, findings presented herein identify SPATS2L, KCTD18, RPL8, HSD17B12, and PEPD of potential importance in controlling fat cell numbers (plasticity), the size of body fat, and diabetes risk.


Assuntos
Diabetes Mellitus Tipo 2 , Estudo de Associação Genômica Ampla , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Contagem de Células , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Loci Gênicos , Humanos , Polimorfismo de Nucleotídeo Único
4.
Surg Obes Relat Dis ; 18(6): 683-692, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35337765

RESUMO

BACKGROUND: There are few long-term mechanistic studies in adipose tissue that investigate the metabolic effects of bariatric surgery. Changes in lipogenesis may be involved in long-term weight development. OBJECTIVES: To investigate the long-term effect of bariatric surgery on lipogenesis in abdominal fat cells and whether surgical treatment could induce an epigenetic memory that would maintain improved lipogenesis in spite of body weight relapse. SETTING: Karolinska University Hospital in Stockholm County, Sweden. METHODS: A total of 22 women with obesity living in the Stockholm area were examined before, 2, 5, and 10 years after bariatric surgery. Abdominal adipose tissue biopsies were obtained. Fat cells were isolated and spontaneous and insulin stimulated glucose incorporation into lipids were assayed. CpG-methylation profiling was performed on adipocytes using the Infinium EPIC BeadChips. RESULTS: Bariatric surgery was associated with improvement in adipocyte spontaneous and insulin stimulated lipogenesis, which was maintained despite some later weight regain (29 % of initial weight loss). There was also an increase in fat cell size between 2- and 10-year follow-up, albeit not to presurgery levels. There were 7729 differentially methylated CpG sites (DMS) at 2 years that showed no sign of return to baseline at either 5 or 10 years. Merging results with expression profiles identified 1259 genes with DMS which showed early response or continual change in expression in one direction after surgery. Upregulated genes with DMS were enriched in gene sets linked to cellular response to insulin stimulus (e.g., IRS1, IRS2, PDE3B, and AKT2) and regulation of lipid metabolic processes. CONCLUSION: Bariatric surgery leads to long-term improvement of lipogenesis and insulin responsiveness in subcutaneous adipocytes in women in spite of some partial body weight regain postoperatively. This may to some extent be explained by epigenetic modifications of fat cell function.


Assuntos
Cirurgia Bariátrica , Adipócitos/patologia , Estudos de Coortes , Feminino , Humanos , Insulina/metabolismo , Estudos Longitudinais , Recidiva , Aumento de Peso , Redução de Peso
5.
Int J Obes (Lond) ; 45(12): 2675-2678, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34321614

RESUMO

Insulin resistance of glucose utilization is fully restored following BMI normalization after bariatric surgery. We investigated if this also pertains to insulin-induced effects on fatty acid handling. Forty-three women with obesity (OB) were investigated before and 2 years after Roux-en-Y gastric by-pass when BMI was <30 kg/m2 (PO) and compared with 26 never obese women (NO). The Adipo-IR index was used as measure of insulin antilipolytic sensitivity. Changes (delta) in circulating glycerol and fatty acid levels during hyperinsulinemic euglycemic clamp represented the insulin maximum antilipolytic effect. Overall fatty acid utilization was reflected by delta fatty acids minus 3 × delta glycerol. Adipo-IR was higher in OB than in NO and PO (p < 0.0001), the latter two groups having similar values. Insulin lowered glycerol levels by about 70% in all groups, but delta glycerol was 30% larger in PO than in NO (p = 0.04). Delta adds and adds utilization were similar in all groups. We conclude that women with obesity, whose BMI is normalized after bariatric surgery, have improved maximum in vivo antilipolytic effect of insulin above expected in absolute but not relative terms as regards glycerol changes, while the handling of circulating fatty acids is changed to the normal state.


Assuntos
Cirurgia Bariátrica/efeitos adversos , Insulina/efeitos adversos , Lipólise/efeitos dos fármacos , Adulto , Cirurgia Bariátrica/estatística & dados numéricos , Glicemia/análise , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Feminino , Humanos , Hiperglicemia/tratamento farmacológico , Hiperglicemia/fisiopatologia , Insulina/uso terapêutico , Lipólise/fisiologia , Estudos Longitudinais , Pessoa de Meia-Idade
6.
Endocrinology ; 162(7)2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33963396

RESUMO

CONTEXT: Healthy hyperplasic (many but smaller fat cells) white adipose tissue (WAT) expansion is mediated by recruitment, proliferation and/or differentiation of new fat cells. This process (adipogenesis) is controlled by transcriptional programs that have been mostly identified in rodents. OBJECTIVE: A systemic investigation of adipogenic human transcription factors (TFs) that are relevant for metabolic conditions has not been revealed previously. METHODS: TFs regulated in WAT by obesity, adipose morphology, cancer cachexia, and insulin resistance were selected from microarrays. Their role in differentiation of human adipose tissue-derived stem cells (hASC) was investigated by RNA interference (RNAi) screen. Lipid accumulation, cell number, and lipolysis were measured for all screened factors (148 TFs). RNA (RNAseq), protein (Western blot) expression, insulin, and catecholamine responsiveness were examined in hASC following siRNA treatment of selected target TFs. RESULTS: Analysis of TFs regulated by metabolic conditions in human WAT revealed that many of them belong to adipogenesis-regulating pathways. The RNAi screen identified 39 genes that affected fat cell differentiation in vitro, where 11 genes were novel. Of the latter JARID2 stood out as being necessary for formation of healthy fat cell metabolic phenotype by regulating expression of multiple fat cell phenotype-specific genes. CONCLUSION: This comprehensive RNAi screening in hASC suggests that a large proportion of WAT TFs that are impacted by metabolic conditions might be important for hyperplastic adipose tissue expansion. The screen also identified JARID2 as a novel TF essential for the development of functional adipocytes.


Assuntos
Adipócitos/metabolismo , Adipogenia/genética , Complexo Repressor Polycomb 2/genética , Interferência de RNA/fisiologia , Fatores de Transcrição/análise , Fatores de Transcrição/genética , Adipócitos/química , Adipócitos/patologia , Tecido Adiposo Branco/química , Tecido Adiposo Branco/patologia , Adolescente , Sequência de Bases , Diferenciação Celular/genética , Células Cultivadas , Feminino , Neoplasias Gastrointestinais , Regulação da Expressão Gênica , Humanos , Hiperplasia/genética , Resistência à Insulina/genética , Masculino , Obesidade/genética , Complexo Repressor Polycomb 2/fisiologia , Células-Tronco/química , Fatores de Transcrição/fisiologia
7.
Diabetes ; 70(7): 1486-1497, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33863803

RESUMO

Selective hepatic insulin resistance is a feature of obesity and type 2 diabetes. Whether similar mechanisms operate in white adipose tissue (WAT) of those with obesity and to what extent these are normalized by weight loss are unknown. We determined insulin sensitivity by hyperinsulinemic euglycemic clamp and insulin response in subcutaneous WAT by RNA sequencing in 23 women with obesity before and 2 years after bariatric surgery. To control for effects of surgery, women postsurgery were matched to never-obese women. Multidimensional analyses of 138 samples allowed us to classify the effects of insulin into three distinct expression responses: a common set was present in all three groups and included genes encoding several lipid/cholesterol biosynthesis enzymes; a set of obesity-attenuated genes linked to tissue remodeling and protein translation was selectively regulated in the two nonobese states; and several postobesity-enriched genes encoding proteins involved in, for example, one-carbon metabolism were only responsive to insulin in the women who had lost weight. Altogether, human WAT displays a selective insulin response in the obese state, where most genes are normalized by weight loss. This comprehensive atlas provides insights into the transcriptional effects of insulin in WAT and may identify targets to improve insulin action.


Assuntos
Tecido Adiposo Branco/metabolismo , Resistência à Insulina , Obesidade/metabolismo , Feminino , Humanos , Metabolismo dos Lipídeos
8.
Nat Rev Endocrinol ; 17(5): 276-295, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33627836

RESUMO

In mammals, the white adipocyte is a cell type that is specialized for storage of energy (in the form of triacylglycerols) and for energy mobilization (as fatty acids). White adipocyte metabolism confers an essential role to adipose tissue in whole-body homeostasis. Dysfunction in white adipocyte metabolism is a cardinal event in the development of insulin resistance and associated disorders. This Review focuses on our current understanding of lipid and glucose metabolic pathways in the white adipocyte. We survey recent advances in humans on the importance of adipocyte hypertrophy and on the in vivo turnover of adipocytes and stored lipids. At the molecular level, the identification of novel regulators and of the interplay between metabolic pathways explains the fine-tuning between the anabolic and catabolic fates of fatty acids and glucose in different physiological states. We also examine the metabolic alterations involved in the genesis of obesity-associated metabolic disorders, lipodystrophic states, cancers and cancer-associated cachexia. New challenges include defining the heterogeneity of white adipocytes in different anatomical locations throughout the lifespan and investigating the importance of rhythmic processes. Targeting white fat metabolism offers opportunities for improved patient stratification and a wide, yet unexploited, range of therapeutic opportunities.


Assuntos
Adipócitos Brancos/metabolismo , Gerenciamento Clínico , Metabolismo dos Lipídeos/fisiologia , Obesidade/metabolismo , Animais , Homeostase , Humanos , Obesidade/terapia
9.
Diabetes ; 70(3): 680-695, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33408126

RESUMO

Elucidation of mechanisms that govern lipid storage, oxidative stress, and insulin resistance may lead to improved therapeutic options for type 2 diabetes and other obesity-related diseases. Here, we find that adipose expression of the small neutral amino acid transporter SLC7A10, also known as alanine-serine-cysteine transporter-1 (ASC-1), shows strong inverse correlates with visceral adiposity, insulin resistance, and adipocyte hypertrophy across multiple cohorts. Concordantly, loss of Slc7a10 function in zebrafish in vivo accelerates diet-induced body weight gain and adipocyte enlargement. Mechanistically, SLC7A10 inhibition in human and murine adipocytes decreases adipocyte serine uptake and total glutathione levels and promotes reactive oxygen species (ROS) generation. Conversely, SLC7A10 overexpression decreases ROS generation and increases mitochondrial respiratory capacity. RNA sequencing revealed consistent changes in gene expression between human adipocytes and zebrafish visceral adipose tissue following loss of SLC7A10, e.g., upregulation of SCD (lipid storage) and downregulation of CPT1A (lipid oxidation). Interestingly, ROS scavenger reduced lipid accumulation and attenuated the lipid-storing effect of SLC7A10 inhibition. These data uncover adipocyte SLC7A10 as a novel important regulator of adipocyte resilience to nutrient and oxidative stress, in part by enhancing glutathione levels and mitochondrial respiration, conducive to decreased ROS generation, lipid accumulation, adipocyte hypertrophy, insulin resistance, and type 2 diabetes.


Assuntos
Adipócitos/metabolismo , Sistema y+ de Transporte de Aminoácidos/metabolismo , Obesidade/metabolismo , Obesidade/fisiopatologia , Células 3T3-L1 , Sistema y+ de Transporte de Aminoácidos/genética , Animais , Western Blotting , Diabetes Mellitus Tipo 2/metabolismo , Genótipo , Glutationa/metabolismo , Humanos , Resistência à Insulina/fisiologia , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Análise de Sequência de RNA , Peixe-Zebra
10.
Cell Metab ; 32(1): 1-3, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32589948

RESUMO

Gao et al. report that the observed reduction in adipose lipolysis with age in women could be explained by an upregulation of the catecholamine-degradation pathway in subcutaneous adipocytes. However, in contrast to findings in mice, these pathways are enriched in adipocytes and not in immune cells, suggesting species-specific differences in aging mechanisms.


Assuntos
Inflamassomos , Lipólise , Adipócitos , Tecido Adiposo/metabolismo , Envelhecimento , Animais , Catecolaminas/metabolismo , Humanos , Inflamassomos/metabolismo , Macrófagos , Camundongos , Norepinefrina
11.
Cells ; 9(5)2020 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-32349335

RESUMO

An increased adipocyte size relative to the size of fat depots, also denoted hypertrophic adipose morphology, is a strong risk factor for the future development of insulin resistance and type 2 diabetes. The regulation of adipose morphology is poorly understood. We set out to identify genetic loci associated with adipose morphology and functionally evaluate candidate genes for impact on adipocyte development. We performed a genome-wide association study (GWAS) in the unique GENetics of Adipocyte Lipolysis (GENiAL) cohort comprising 948 participants who have undergone abdominal subcutaneous adipose biopsy with a determination of average adipose volume and morphology. The GWAS identified 31 genetic loci displaying suggestive association with adipose morphology. Functional evaluation of candidate genes by small interfering RNAs (siRNA)-mediated knockdown in adipose-derived precursor cells identified six genes controlling adipocyte renewal and differentiation, and thus of potential importance for adipose hypertrophy. In conclusion, genetic and functional studies implicate a regulatory role for ATL2, ARHGEF10, CYP1B1, TMEM200A, C17orf51, and L3MBTL3 in adipose morphology by their impact on adipogenesis.


Assuntos
Adipócitos/citologia , Diabetes Mellitus Tipo 2/genética , Obesidade/genética , Adipócitos/fisiologia , Adipogenia/genética , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Adiposidade , Adulto , Diferenciação Celular , Estudos de Coortes , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Estudo de Associação Genômica Ampla/métodos , Humanos , Insulina/metabolismo , Resistência à Insulina/fisiologia , Lipólise/fisiologia , Masculino , Pessoa de Meia-Idade , Gordura Subcutânea
12.
Nutrients ; 11(12)2019 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-31816875

RESUMO

A healthy dietary pattern is associated with a lower risk of metabolic syndrome (MetS) and reduced inflammation. To explore this at the molecular level, we investigated the effect of a Nordic diet (ND) on changes in the gene expression profiles of inflammatory and lipid-related genes in peripheral blood mononuclear cells (PBMCs) of individuals with MetS. We hypothesized that the intake of an ND compared to a control diet (CD) would alter the expression of inflammatory genes and genes involved in lipid metabolism. The individuals with MetS underwent an 18/24-week randomized intervention to compare a ND with a CD. Eighty-eight participants (66% women) were included in this sub-study of the larger SYSDIET study. Fasting PBMCs were collected before and after the intervention and changes in gene expression levels were measured using TaqMan Array Micro Fluidic Cards. Forty-eight pre-determined inflammatory and lipid related gene transcripts were analyzed. The expression level of the gene tumor necrosis factor (TNF) receptor superfamily member 1A (TNFRSF1A) was down-regulated (p = 0.004), whereas the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) subunit, RELA proto-oncogene, was up-regulated (p = 0.016) in the ND group compared to the CD group. In conclusion, intake of an ND in individuals with the MetS may affect immune function.


Assuntos
Dietoterapia , Leucócitos Mononucleares/efeitos dos fármacos , Síndrome Metabólica/dietoterapia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fator de Transcrição RelA/metabolismo , Adulto , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Proto-Oncogene Mas , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Fator de Transcrição RelA/genética , Transcriptoma
13.
Metabolism ; 101: 153999, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31672447

RESUMO

BACKGROUND: Adipose tissue plays a crucial role in diet- and obesity-related insulin resistance, with implications for several metabolic diseases. Identification of novel target genes and mechanisms that regulate adipocyte function could lead to improved treatment strategies. RND3 (RhoE/Rho8), a Rho-related GTP-binding protein that inhibits Rho kinase (ROCK) signaling, has been linked to diverse diseases such as apoptotic cardiomyopathy, heart failure, cancer and type 2 diabetes, in part by regulating cytoskeleton dynamics and insulin-mediated glucose uptake. RESULTS: We here investigated the expression of RND3 in adipose tissue in human obesity, and discovered a role for RND3 in regulating adipocyte metabolism. In cross-sectional and prospective studies, we observed 5-fold increased adipocyte levels of RND3 mRNA in obesity, reduced levels after surgery-induced weight loss, and positive correlations of RND3 mRNA with adipocyte size and surrogate measures of insulin resistance (HOMA2-IR and circulating triglyceride/high-density lipoprotein cholesterol (TAG/HDL-C) ratio). By screening for RND3-dependent gene expression following siRNA-mediated RND3 knockdown in differentiating human adipocytes, we found downregulation of inflammatory genes and upregulation of genes related to adipocyte ipolysis and insulin signaling. Treatment of adipocytes with tumor necrosis factor alpha (TNFα), lipopolysaccharide (LPS), hypoxia or cAMP analogs increased RND3 mRNA levels 1.5-2-fold. Functional assays in primary human adipocytes confirmed that RND3 knockdown reduces cAMP- and isoproterenol-induced lipolysis, which were mimicked by treating cells with ROCK inhibitor. This effect could partly be explained by reduced protein expression of adipose triglyceride lipase (ATGL) and phosphorylated hormone-sensitive lipase (HSL). CONCLUSION: We here uncovered a novel differential expression of adipose RND3 in obesity and insulin resistance, which may at least partly depend on a causal effect of RND3 on adipocyte lipolysis.


Assuntos
Adipócitos/metabolismo , Lipólise/efeitos dos fármacos , Proteínas rho de Ligação ao GTP/fisiologia , Animais , Células Cultivadas , Estudos Transversais , Regulação da Expressão Gênica , Humanos , Resistência à Insulina , Obesidade/metabolismo , Estudos Prospectivos , RNA Mensageiro/metabolismo , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo
14.
J Clin Endocrinol Metab ; 104(10): 4552-4562, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31132124

RESUMO

OBJECTIVE: Although IL-10 is generally considered as an anti-inflammatory cytokine, it was recently shown to have detrimental effects on insulin sensitivity and fat cell metabolism in rodents. Whether this also pertains to human white adipose tissue (hWAT) is unclear. We therefore determined the main cellular source and effects of IL-10 on human adipocytes and hWAT-resident immune cells and its link to insulin resistance. METHODS: Associations between hWAT IL-10 production and metabolic parameters were investigated in 216 participants with large interindividual variations in body mass index and insulin sensitivity. Adipose cells expressing or secreting IL-10 and the cognate IL-10 receptor α (IL10RA) were identified by flow cytometry sorting. Effects on adipogenesis, lipolysis, and inflammatory/metabolic gene expression were measured in two human primary adipocyte models. Secretion of inflammatory cytokines was investigated in cultures of IL-10-treated hWAT macrophages and leukocytes by Luminex analysis (Luminex Corp.). RESULTS: IL-10 gene expression and protein secretion in hWAT correlated positively with body mass index (BMI) and homeostasis model assessment-insulin resistance (HOMA-IR). Gene expression analyses in mature fat cells and flow cytometry-sorted hWAT-resident adipocyte progenitors, macrophages, and leukocytes demonstrated that the expression of IL-10 and the IL10RA were significantly enriched in proinflammatory M1 macrophages. In contrast to murine data, functional studies showed that recombinant IL-10 had no effect on adipocyte phenotype. In hWAT-derived macrophages and leukocytes, it induced an anti-inflammatory profile. CONCLUSION: In hWAT, IL-10 is upregulated in proinflammatory macrophages of obese and insulin-resistant persons. However, in contrast to findings in mice, IL-10 does not directly affect human adipocyte function.


Assuntos
Tecido Adiposo Branco/metabolismo , Resistência à Insulina , Interleucina-10/metabolismo , Macrófagos/metabolismo , Adipócitos Brancos/efeitos dos fármacos , Adipócitos Brancos/metabolismo , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Tecido Adiposo/metabolismo , Adulto , Estudos de Casos e Controles , Células Cultivadas , Feminino , Citometria de Fluxo , Expressão Gênica , Humanos , Técnicas In Vitro , Inflamação/genética , Interleucina-10/farmacologia , Subunidade alfa de Receptor de Interleucina-10/metabolismo , Gordura Intra-Abdominal , Lipólise/genética , Células-Tronco Mesenquimais/efeitos dos fármacos , Pessoa de Meia-Idade , Obesidade Metabolicamente Benigna , RNA Mensageiro , Gordura Subcutânea , Células THP-1 , Adulto Jovem
15.
Am J Clin Nutr ; 109(6): 1499-1510, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30869115

RESUMO

BACKGROUND: The adipose tissue (AT) is a secretory organ producing a wide variety of factors that participate in the genesis of metabolic disorders linked to excess fat mass. Weight loss improves obesity-related disorders. OBJECTIVES: Transcriptomic studies on human AT, and a combination of analyses of transcriptome and proteome profiling of conditioned media from adipocytes and stromal cells isolated from human AT, have led to the identification of apolipoprotein M (apoM) as a putative adipokine. We aimed to validate apoM as novel adipokine, investigate the relation of AT APOM expression with metabolic syndrome and insulin sensitivity, and study the regulation of its expression in AT and secretion during calorie restriction-induced weight loss. METHODS: We examined APOM mRNA level and secretion in AT from 485 individuals enrolled in 5 independent clinical trials, and in vitro in human multipotent adipose-derived stem cell adipocytes. APOM expression and secretion were measured during dieting. RESULTS: APOM was expressed in human subcutaneous and visceral AT, mainly by adipocytes. ApoM was released into circulation from AT, and plasma apoM concentrations correlate with AT APOM mRNA levels. In AT, APOM expression inversely correlated with adipocyte size, was lower in obese compared to lean individuals, and reduced in subjects with metabolic syndrome and type 2 diabetes. Regardless of fat depot, there was a positive relation between AT APOM expression and systemic insulin sensitivity, independently of fat mass and plasma HDL cholesterol. In human multipotent adipose-derived stem cell adipocytes, APOM expression was enhanced by insulin-sensitizing peroxisome proliferator-activated receptor agonists and inhibited by tumor necrosis factor α, a cytokine that causes insulin resistance. In obese individuals, calorie restriction increased AT APOM expression and secretion. CONCLUSIONS: ApoM is a novel adipokine, the expression of which is a hallmark of healthy AT and is upregulated by calorie restriction. AT apoM deserves further investigation as a potential biomarker of risk for diabetes and cardiovascular diseases.


Assuntos
Adipocinas/genética , Apolipoproteínas M/genética , Obesidade/dietoterapia , Obesidade/genética , Adipócitos/metabolismo , Adipocinas/metabolismo , Apolipoproteínas M/metabolismo , Restrição Calórica , Ensaios Clínicos como Assunto , Estudos de Coortes , Estudos Transversais , Feminino , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Obesidade/metabolismo
16.
Nat Commun ; 9(1): 5068, 2018 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-30498206

RESUMO

Methylation patterns of circulating cell-free DNA (cfDNA) contain rich information about recent cell death events in the body. Here, we present an approach for unbiased determination of the tissue origins of cfDNA, using a reference methylation atlas of 25 human tissues and cell types. The method is validated using in silico simulations as well as in vitro mixes of DNA from different tissue sources at known proportions. We show that plasma cfDNA of healthy donors originates from white blood cells (55%), erythrocyte progenitors (30%), vascular endothelial cells (10%) and hepatocytes (1%). Deconvolution of cfDNA from patients reveals tissue contributions that agree with clinical findings in sepsis, islet transplantation, cancer of the colon, lung, breast and prostate, and cancer of unknown primary. We propose a procedure which can be easily adapted to study the cellular contributors to cfDNA in many settings, opening a broad window into healthy and pathologic human tissue dynamics.


Assuntos
Ácidos Nucleicos Livres/genética , Algoritmos , Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Células Cultivadas , Neoplasias do Colo/genética , Ilhas de CpG/genética , Metilação de DNA/genética , Células Endoteliais/metabolismo , Eritrócitos/metabolismo , Hepatócitos/metabolismo , Humanos , Leucócitos/metabolismo , Neoplasias Pulmonares/genética , Regiões Promotoras Genéticas/genética , Sepse/genética
17.
Artigo em Inglês | MEDLINE | ID: mdl-29991030

RESUMO

Lipid storage and release from fat cells in adipose tissue are key factors in the regulation of the energy balance. During infancy and adolescence, adipose tissue is growing by a combination of increase in fat cell size (to a lesser extent) and (above all) the number of these cells. In adults, fat cell number is constant over time in spite of a large turnover (about 10% of the fat cells per year) when body weight is stable. A decrease in body weight only changes fat cell size (becoming smaller), whereas an increase in body weight causes elevation of both fat cell size and number in adults. An important source of renewal of fat cells during the entire life span is the bone marrow. This is most apparent in obesity when ∼20% of all fat cells are derived from the bone marrow. Fat cell turnover is also important for the size of fat cells. Low turnover may cause large fat cells which, in turn, is linked to cardiovascular disease and type 2 diabetes. There is also a rapid turnover of fat cell lipids, which constitute a single active pool and are renewed about 6 times during the life span of individual fat cells. Overweight and obesity are associated with decreased lipid turnover due to high input in combination with low output of lipids from the fat cells. Low fat cell lipid turnover is associated with insulin resistance and dyslipidemia. Thus, changes in the turnover of fat cells and their lipid content are important for the development of adipose tissue mass and its cellularity (fat cell size and number) and, in turn, for metabolic disturbances.


Assuntos
Tecido Adiposo/crescimento & desenvolvimento , Adipócitos Brancos/química , Adipócitos Brancos/patologia , Adipócitos Brancos/fisiologia , Tecido Adiposo/citologia , Adolescente , Peso Corporal/fisiologia , Células da Medula Óssea/fisiologia , Doenças Cardiovasculares/patologia , Contagem de Células , Tamanho Celular , Diabetes Mellitus Tipo 2/patologia , Dislipidemias/metabolismo , Humanos , Hiperplasia , Hipertrofia , Lactente , Resistência à Insulina/fisiologia , Metabolismo dos Lipídeos , Lipídeos/análise , Obesidade/patologia , Sobrepeso/patologia
18.
Surg Obes Relat Dis ; 14(7): 936-942, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29705562

RESUMO

BACKGROUND: Bariatric surgery such as Roux-en-Y gastric bypass (RYGB) remains the most effective treatment of obesity and associated co-morbidities. Body fat distribution associates with metabolic function. OBJECTIVE: To investigate if preoperative body fat mass and distribution measured by dual-energy x-ray absorptiometry (DXA) predict weight loss and metabolic outcome after RYGB, and to compare predictive value of DXA with simple anthropometric measures. SETTING: Four Swedish hospitals within the Stockholm area. METHODS: Two hundred fifteen women scheduled for RYGB were included. Evaluations before and 2 years after RYGB included determination of insulin sensitivity by the homeostatic model assessment of insulin resistance, blood pressure, plasma lipids, and anthropometric measures, such as waist-to-hip-ratio and fat percentage estimated by formula. Body fat mass and distribution were determined by DXA. RESULTS: Follow-up rate was 77.2% (n = 166). All clinical, anthropometric, and DXA measures were improved/reduced postsurgery (all P<.0001). Android/gynoid fat mass ratio and waist-to-hip-ratio predicted improved homeostatic model assessment of insulin resistance (P = .0028 and .0014), independently of body mass index and age. Body fat percentage, measured by DXA or estimated by formula, predicted percent weight loss (P<.0001 and .0083). Body mass index predicted percent weight loss and percent excess body mass index lost (P = .0022 and<.0001). DXA and anthropometric measures performed equally as predictors, except for DXA measured fat percentage that was slightly better than formula estimated. CONCLUSION: DXA provided predictive values similar to those by basic anthropometric measures, suggesting a limited additional value of preoperative DXA to predict metabolic improvement and weight loss after RYGB in women.


Assuntos
Anastomose em-Y de Roux/métodos , Distribuição da Gordura Corporal/métodos , Derivação Gástrica/métodos , Obesidade Mórbida/cirurgia , Redução de Peso/fisiologia , Absorciometria de Fóton/métodos , Adulto , Índice de Massa Corporal , Estudos de Coortes , Feminino , Seguimentos , Humanos , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Cuidados Pré-Operatórios/métodos , Suécia , Fatores de Tempo , Resultado do Tratamento
19.
Int J Obes (Lond) ; 42(12): 2047-2056, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29453465

RESUMO

BACKGROUND: Adipokines are peptides secreted from white adipose tissue (WAT), which have been linked to WAT dysfunction and metabolic complications of obesity. We set out to identify novel adipokines in subcutaneous WAT (sWAT) linked to insulin resistance (IR). METHODS: Gene expression was determined by microarray and qPCR in obese and non-obese subjects with varying degree of IR. WAT-secreted and circulating protein levels were measured by ELISA. RESULTS: In sWAT of 80 obese women discordant for IR, 44 genes encoding potential adipose-secreted proteins were differentially expressed. Among these, merely two proteins, S100A4 and MXRA5 were released from sWAT in a time-dependent manner (criterion for true adipokines) but only the circulating levels of S100A4 were higher in IR. In two additional cohorts (n = 29 and n = 56), sWAT S100A4 secretion was positively and BMI-independently associated with IR (determined by clamp or HOMA-IR), ATP-III risk score and adipocyte size (hypertrophy). In non-obese (n = 20) and obese subjects before and after bariatric surgery (n = 21), circulating and sWAT-secreted levels were highest in the obese and normalized following weight loss. Serum S100A4 concentrations were higher in subjects with type 2 diabetes. S100A4 sWAT expression associated positively with genes involved in inflammation/extracellular matrix formation and inversely with genes in metabolic pathways. Although S100A4 was expressed in both stromal cells and adipocytes, only the expression in adipocytes associated with BMI. CONCLUSIONS: S100A4 is a novel adipokine associated with IR and sWAT inflammation/adipocyte hypertrophy independently of BMI. Its value as a circulating marker for dysfunctional WAT and IR needs to be validated in larger cohorts.


Assuntos
Adipocinas/metabolismo , Tecido Adiposo Branco/metabolismo , Resistência à Insulina/fisiologia , Obesidade/metabolismo , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Adipocinas/sangue , Tecido Adiposo Branco/química , Adulto , Biomarcadores , Estudos de Coortes , Feminino , Humanos , Inflamação/metabolismo , Masculino , Pessoa de Meia-Idade , Obesidade/sangue , Obesidade/epidemiologia , Proteína A4 de Ligação a Cálcio da Família S100/sangue
20.
Sci Rep ; 8(1): 1097, 2018 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-29348496

RESUMO

Robust associations between low plasma level of natriuretic peptides (NP) and increased risk of type 2 diabetes (T2D) have been recently reported in humans. Adipose tissue (AT) is a known target of NP. However it is unknown whether NP signalling in human AT relates to insulin sensitivity and modulates glucose metabolism. We here show in two European cohorts that the NP receptor guanylyl cyclase-A (GC-A) expression in subcutaneous AT was down-regulated as a function of obesity grade while adipose NP clearance receptor (NPRC) was up-regulated. Adipose GC-A mRNA level was down-regulated in prediabetes and T2D, and negatively correlated with HOMA-IR and fasting blood glucose. We show for the first time that NP promote glucose uptake in a dose-dependent manner. This effect is reduced in adipocytes of obese individuals. NP activate mammalian target of rapamycin complex 1/2 (mTORC1/2) and Akt signalling. These effects were totally abrogated by inhibition of cGMP-dependent protein kinase and mTORC1/2 by rapamycin. We further show that NP treatment favoured glucose oxidation and de novo lipogenesis independently of significant gene regulation. Collectively, our data support a role for NP in blood glucose control and insulin sensitivity by increasing glucose uptake in human adipocytes. This effect is partly blunted in obesity.


Assuntos
Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , GMP Cíclico/metabolismo , Glucose/metabolismo , Peptídeos Natriuréticos/farmacologia , Tecido Adiposo/metabolismo , Biomarcadores , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Regulação da Expressão Gênica , Humanos , Resistência à Insulina , Modelos Biológicos , Obesidade/genética , Obesidade/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptores do Fator Natriurético Atrial/genética , Receptores do Fator Natriurético Atrial/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA